Dysregulated Lipid Metabolism as a Central Driver of Atherosclerotic Plaque Pathology
Abstract
1. Introduction
2. Hepatic Synthesis, Composition, and Transport of Circulating Lipoproteins
3. Lipid-Dependent Aortic Endothelial Dysfunction and Monocyte Entry
4. Macrophage Scavenging and Metabolism of Lipids
5. Fatty Acids as Signaling Molecules
6. Other Lipid Species as Drivers of Atherosclerosis
7. Lipids as Drivers of Non-Apoptotic Cell Death
8. Lipid Therapies in Atherosclerotic Cardiovascular Disease
9. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
AA | Arachidonic acid |
ABCA1/G1/G5/G8 | Adenosine triphosphate-binding cassette transporter A1/G1/G5/G8 |
ACAT1 | Acetyl-CoA acetyltransferase 1 |
ACLY | ATP citrate lyase |
agLDL | Aggregated LDL |
ANGPTL3/4/8 | Angiopoietin-like 3/4/8 |
Apo-A1/B100/C3/(a) | Apolipoprotein A1/B100/C3/(a) |
ASCVD | Atherosclerotic cardiovascular disease |
ATF6 | Activating transcription factor 6 |
ATP | Adenosine triphosphate |
CAD | Coronary artery disease |
CCL2/MCP-1 | Chemokine (C-C motif) ligand 2/Monocyte chemoattractant protein 1 |
CCR2 | C-C chemokine receptor type 2 |
CD36 | Cluster of differentiation 36 |
CETP | Cholesteryl ester transfer protein |
CHOP | CCAAT-enhancer-binding protein homologous protein |
COX | Cyclooxygenase |
CVD | Cardiovascular disease |
CYP7A1 | Cholesterol 7 alpha-hydroxylase |
DHA | Docosahexaenoic acid |
DOCK4 | Dedicator of cytokinesis 4 |
EET | Epoxyeicosatetraenoic acid |
EndMT | Endothelial-to-mesenchymal transition |
EPA | Eicosapentaenoic acid |
ER | Endoplasmic reticulum |
FH | Familial hypercholesterolemia |
GPCR | G protein-coupled receptor |
HETE | Hydroeicosatetraenoic acid |
HDL | High-density lipoprotein |
HMOX1 | Heme oxygenase 1 |
ICAM-1 | Intercellular cell adhesion molecule 1 |
IDL | Intermediate-density lipoprotein |
IL-1β/8 | Interleukin-1β/8 |
IRE1 | Inositol-requiring enzyme 1 |
LAL | Lysosomal acid lipase |
LDL | Low-density lipoprotein |
LDLR | Low-density lipoprotein receptor |
LOX-1/OLR1 | Lectin-like oxidized low-density lipoprotein receptor 1/OxLDL receptor 1 |
Lp(a) | Lipoprotein(a) |
LPL | Lipoprotein lipase |
LXRα/β | Liver x receptor α/β |
M-CSF/CSF1 | Macrophage-colony stimulating factor/Colony-stimulating factor 1 |
MACE | Major adverse cardiovascular events |
MLKL | Mixed lineage kinase domain-like protein |
NF-κB | Nuclear factor kappa-light-chain-enhancer of activated B cells |
NLRP3 | NOD-, LRR- and pyrin domain-containing protein 3 |
oxLDL | Oxidized low-density lipoprotein |
oxPAPC | Oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine |
oxPL | Oxidized phospholipids |
PCSK9 | Proprotein convertase subtilisin/kexin type 9 |
PERK | PKR-like eukaryotic initiation factor 2A kinase |
POVPC | 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine |
PUFA | Polyunsaturated fatty acid |
RCT | Reverse cholesterol transport |
rHDL | Recombinant HDL |
RIPK1/3 | Receptor-interacting serine/threonine-protein kinase 1/3 |
RvD1/E1 | Resolvin D1/E1 |
SPM | Specialized pro-resolving mediators |
SR-A/B1 | Scavenger receptor class A/class B member 1 |
TLR2/4/6 | Toll-like receptor 2/4/6 |
TRL | Triglyceride-rich lipoprotein |
UPR | Unfolded protein response |
VCAM-1 | Vascular cell adhesion molecule 1 |
VLDL | Very low-density lipoprotein |
References
- Ahmed, S.; Shah, P.; Ahmed, O. Biochemistry, Lipids. In StatPearls; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
- Feingold, K.R. Introduction to Lipids and Lipoproteins. In Endotext; Feingold, K.R., Ahmed, S.F., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
- Yoon, H.; Shaw, J.L.; Haigis, M.C.; Greka, A. Lipid metabolism in sickness and in health: Emerging regulators of lipotoxicity. Mol. Cell 2021, 81, 3708–3730. [Google Scholar] [CrossRef]
- World Health Organization. Global Health Estimates: Life Expectancy and Leading Causes of Death and Disability. Available online: https://www.who.int/data/gho/data/themes/mortality-and-global-health-estimates (accessed on 7 August 2025).
- Nowbar, A.N.; Gitto, M.; Howard, J.P.; Francis, D.P.; Al-Lamee, R. Mortality From Ischemic Heart Disease. Circ. Cardiovasc. Qual. Outcomes 2019, 12, e005375. [Google Scholar] [CrossRef]
- Martin, S.S.; Aday, A.W.; Allen, N.B.; Almarzooq, Z.I.; Anderson, C.A.M.; Arora, P.; Avery, C.L.; Baker-Smith, C.M.; Bansal, N.; Beaton, A.Z.; et al. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025, 151, e41–e660. [Google Scholar] [CrossRef] [PubMed]
- Stary, H.C.; Chandler, A.B.; Dinsmore, R.E.; Fuster, V.; Glagov, S.; Insull, W., Jr.; Rosenfeld, M.E.; Schwartz, C.J.; Wagner, W.D.; Wissler, R.W. A definition of advanced types of atherosclerotic lesions and a histological classification of atherosclerosis. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation 1995, 92, 1355–1374. [Google Scholar] [CrossRef]
- Stary, H.C. Natural history and histological classification of atherosclerotic lesions: An update. Arter. Thromb. Vasc. Biol. 2000, 20, 1177–1178. [Google Scholar] [CrossRef] [PubMed]
- Libby, P.; Pasterkamp, G.; Crea, F.; Jang, I.K. Reassessing the Mechanisms of Acute Coronary Syndromes. Circ. Res. 2019, 124, 150–160. [Google Scholar] [CrossRef] [PubMed]
- Vergallo, R.; Park, S.J.; Stone, G.W.; Erlinge, D.; Porto, I.; Waksman, R.; Mintz, G.S.; D’Ascenzo, F.; Seitun, S.; Saba, L.; et al. Vulnerable or High-Risk Plaque: A JACC: Cardiovascular Imaging Position Statement. JACC Cardiovasc. Imaging 2025, 18, 709–740. [Google Scholar] [CrossRef]
- Arnett, D.K.; Blumenthal, R.S.; Albert, M.A.; Buroker, A.B.; Goldberger, Z.D.; Hahn, E.J.; Himmelfarb, C.D.; Khera, A.; Lloyd-Jones, D.; McEvoy, J.W.; et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019, 140, e563–e595. [Google Scholar] [CrossRef]
- Sorci-Thomas, M.G.; Hegele, R.A.; Remaley, A.T. A Century of Milestones and Breakthroughs Related to Low- and High-Density Lipoproteins. Arter. Thromb. Vasc. Biol. 2024, 44, 7–11. [Google Scholar] [CrossRef]
- Juarez Casso, F.M.; Farzam, K. Biochemistry, Very Low Density Lipoprotein. In StatPearls; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
- Chen, J.; Fang, Z.; Luo, Q.; Wang, X.; Warda, M.; Das, A.; Oldoni, F.; Luo, F. Unlocking the mysteries of VLDL: Exploring its production, intracellular trafficking, and metabolism as therapeutic targets. Lipids Health Dis. 2024, 23, 14. [Google Scholar] [CrossRef]
- Chapman, M.J.; Ginsberg, H.N.; Amarenco, P.; Andreotti, F.; Boren, J.; Catapano, A.L.; Descamps, O.S.; Fisher, E.; Kovanen, P.T.; Kuivenhoven, J.A.; et al. Triglyceride-rich lipoproteins and high-density lipoprotein cholesterol in patients at high risk of cardiovascular disease: Evidence and guidance for management. Eur. Heart J. 2011, 32, 1345–1361. [Google Scholar] [CrossRef]
- Ginsberg, H.N.; Packard, C.J.; Chapman, M.J.; Boren, J.; Aguilar-Salinas, C.A.; Averna, M.; Ference, B.A.; Gaudet, D.; Hegele, R.A.; Kersten, S.; et al. Triglyceride-rich lipoproteins and their remnants: Metabolic insights, role in atherosclerotic cardiovascular disease, and emerging therapeutic strategies-a consensus statement from the European Atherosclerosis Society. Eur. Heart J. 2021, 42, 4791–4806. [Google Scholar] [CrossRef] [PubMed]
- Goldberg, I.J. Lipoprotein lipase and lipolysis: Central roles in lipoprotein metabolism and atherogenesis. J. Lipid Res. 1996, 37, 693–707. [Google Scholar] [CrossRef]
- Gofman, J.W.; Lindgren, F. The role of lipids and lipoproteins in atherosclerosis. Science 1950, 111, 166–171. [Google Scholar] [CrossRef]
- Gofman, J.W.; Jones, H.B.; Lyon, T.P.; Lindgren, F.; Strisower, B.; Colman, D.; Herring, V. Blood lipids and human atherosclerosis. Circulation 1952, 5, 119–134. [Google Scholar] [CrossRef]
- Goldstein, J.L.; Brown, M.S. A century of cholesterol and coronaries: From plaques to genes to statins. Cell 2015, 161, 161–172. [Google Scholar] [CrossRef]
- Goldstein, J.L.; Brown, M.S. The LDL receptor. Arter. Thromb. Vasc. Biol. 2009, 29, 431–438. [Google Scholar] [CrossRef]
- Berndsen, Z.T.; Cassidy, C.K. The structure of apolipoprotein B100 from human low-density lipoprotein. Nature 2025, 638, 836–843. [Google Scholar] [CrossRef] [PubMed]
- Reimund, M.; Dearborn, A.D.; Graziano, G.; Lei, H.; Ciancone, A.M.; Kumar, A.; Holewinski, R.; Neufeld, E.B.; O’Reilly, F.J.; Remaley, A.T.; et al. Structure of apolipoprotein B100 bound to the low-density lipoprotein receptor. Nature 2025, 638, 829–835. [Google Scholar] [CrossRef]
- Brown, M.S.; Goldstein, J.L. A receptor-mediated pathway for cholesterol homeostasis. Science 1986, 232, 34–47. [Google Scholar] [CrossRef] [PubMed]
- Davidson, W.S. The structure of bad cholesterol comes into focus. Nature 2025, 638, 618–620. [Google Scholar] [CrossRef] [PubMed]
- Powers, H.R.; Sahoo, D. SR-B1’s Next Top Model: Structural Perspectives on the Functions of the HDL Receptor. Curr. Atheroscler. Rep. 2022, 24, 277–288. [Google Scholar] [CrossRef]
- Linton, M.F.; Yancey, P.G.; Tao, H.; Davies, S.S. HDL Function and Atherosclerosis: Reactive Dicarbonyls as Promising Targets of Therapy. Circ. Res. 2023, 132, 1521–1545. [Google Scholar] [CrossRef]
- Ouimet, M.; Barrett, T.J.; Fisher, E.A. HDL and Reverse Cholesterol Transport. Circ. Res. 2019, 124, 1505–1518. [Google Scholar] [CrossRef]
- Bae, H.; Jung, S.; Le, J.; Tamburini, I.; Kim, J.; Wang, E.; Song, W.S.; Choi, W.; Jang, K.H.; Kang, T.; et al. Cross-organ metabolite production and consumption in healthy and atherogenic conditions. Cell 2025, 188, 4441–4455.e16. [Google Scholar] [CrossRef]
- Sturm, A.C.; Knowles, J.W.; Gidding, S.S.; Ahmad, Z.S.; Ahmed, C.D.; Ballantyne, C.M.; Baum, S.J.; Bourbon, M.; Carrie, A.; Cuchel, M.; et al. Clinical Genetic Testing for Familial Hypercholesterolemia: JACC Scientific Expert Panel. J. Am. Coll. Cardiol. 2018, 72, 662–680. [Google Scholar] [CrossRef]
- McGowan, M.P.; Hosseini Dehkordi, S.H.; Moriarty, P.M.; Duell, P.B. Diagnosis and Treatment of Heterozygous Familial Hypercholesterolemia. J. Am. Heart Assoc. 2019, 8, e013225. [Google Scholar] [CrossRef]
- Lee, Y.; Siddiqui, W.J. Cholesterol Levels. In StatPearls; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
- Farzam, K.; Zubair, M.; Senthilkumaran, S. Lipoprotein A. In StatPearls; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
- Koschinsky, M.L.; Bajaj, A.; Boffa, M.B.; Dixon, D.L.; Ferdinand, K.C.; Gidding, S.S.; Gill, E.A.; Jacobson, T.A.; Michos, E.D.; Safarova, M.S.; et al. A focused update to the 2019 NLA scientific statement on use of lipoprotein(a) in clinical practice. J Clin Lipidol 2024, 18, e308–e319. [Google Scholar] [CrossRef] [PubMed]
- Tsimikas, S.; Fazio, S.; Ferdinand, K.C.; Ginsberg, H.N.; Koschinsky, M.L.; Marcovina, S.M.; Moriarty, P.M.; Rader, D.J.; Remaley, A.T.; Reyes-Soffer, G.; et al. NHLBI Working Group Recommendations to Reduce Lipoprotein(a)-Mediated Risk of Cardiovascular Disease and Aortic Stenosis. J. Am. Coll. Cardiol. 2018, 71, 177–192. [Google Scholar] [CrossRef] [PubMed]
- Madsen, C.M.; Kamstrup, P.R.; Langsted, A.; Varbo, A.; Nordestgaard, B.G. Lipoprotein(a)-Lowering by 50 mg/dL (105 nmol/L) May Be Needed to Reduce Cardiovascular Disease 20% in Secondary Prevention: A Population-Based Study. Arter. Thromb. Vasc. Biol. 2020, 40, 255–266. [Google Scholar] [CrossRef]
- Ross, R.; Glomset, J.A. The pathogenesis of atherosclerosis (first of two parts). N. Engl. J. Med. 1976, 295, 369–377. [Google Scholar] [CrossRef]
- Ross, R.; Glomset, J.A. The pathogenesis of atherosclerosis (second of two parts). N. Engl. J. Med. 1976, 295, 420–425. [Google Scholar] [CrossRef]
- Ference, B.A.; Braunwald, E.; Catapano, A.L. The LDL cumulative exposure hypothesis: Evidence and practical applications. Nat. Rev. Cardiol. 2024, 21, 701–716. [Google Scholar] [CrossRef] [PubMed]
- Williams, K.J.; Tabas, I. The response-to-retention hypothesis of early atherogenesis. Arter. Thromb. Vasc. Biol. 1995, 15, 551–561. [Google Scholar] [CrossRef] [PubMed]
- Steinberg, D. Atherogenesis in perspective: Hypercholesterolemia and inflammation as partners in crime. Nat. Med. 2002, 8, 1211–1217. [Google Scholar] [CrossRef]
- Cheng, C.K.; Wang, N.; Wang, L.; Huang, Y. Biophysical and Biochemical Roles of Shear Stress on Endothelium: A Revisit and New Insights. Circ. Res. 2025, 136, 752–772. [Google Scholar] [CrossRef] [PubMed]
- Souilhol, C.; Serbanovic-Canic, J.; Fragiadaki, M.; Chico, T.J.; Ridger, V.; Roddie, H.; Evans, P.C. Endothelial responses to shear stress in atherosclerosis: A novel role for developmental genes. Nat. Rev. Cardiol. 2020, 17, 52–63. [Google Scholar] [CrossRef]
- Botts, S.R.; Fish, J.E.; Howe, K.L. Dysfunctional Vascular Endothelium as a Driver of Atherosclerosis: Emerging Insights Into Pathogenesis and Treatment. Front. Pharmacol. 2021, 12, 787541. [Google Scholar] [CrossRef]
- Gimbrone, M.A., Jr.; Garcia-Cardena, G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016, 118, 620–636. [Google Scholar] [CrossRef]
- Souilhol, C.; Harmsen, M.C.; Evans, P.C.; Krenning, G. Endothelial-mesenchymal transition in atherosclerosis. Cardiovasc. Res. 2018, 114, 565–577. [Google Scholar] [CrossRef]
- De Caterina, R.; Libby, P.; Peng, H.B.; Thannickal, V.J.; Rajavashisth, T.B.; Gimbrone, M.A., Jr.; Shin, W.S.; Liao, J.K. Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines. J. Clin. Investig. 1995, 96, 60–68. [Google Scholar] [CrossRef]
- Marshall, H.E.; Stamler, J.S. Nitrosative stress-induced apoptosis through inhibition of NF-kappa B. J. Biol. Chem. 2002, 277, 34223–34228. [Google Scholar] [CrossRef]
- Matthews, J.R.; Botting, C.H.; Panico, M.; Morris, H.R.; Hay, R.T. Inhibition of NF-kappaB DNA binding by nitric oxide. Nucleic Acids Res. 1996, 24, 2236–2242. [Google Scholar] [CrossRef] [PubMed]
- Reynaert, N.L.; Ckless, K.; Korn, S.H.; Vos, N.; Guala, A.S.; Wouters, E.F.; van der Vliet, A.; Janssen-Heininger, Y.M. Nitric oxide represses inhibitory kappaB kinase through S-nitrosylation. Proc. Natl. Acad. Sci. USA 2004, 101, 8945–8950. [Google Scholar] [CrossRef] [PubMed]
- Mundi, S.; Massaro, M.; Scoditti, E.; Carluccio, M.A.; van Hinsbergh, V.W.M.; Iruela-Arispe, M.L.; De Caterina, R. Endothelial permeability, LDL deposition, and cardiovascular risk factors-a review. Cardiovasc. Res. 2018, 114, 35–52. [Google Scholar] [CrossRef]
- Hirata, K.; Akita, H.; Yokoyama, M. Oxidized low density lipoprotein inhibits bradykinin-induced phosphoinositide hydrolysis in cultured bovine aortic endothelial cells. FEBS Lett. 1991, 287, 181–184. [Google Scholar] [CrossRef] [PubMed]
- Inoue, N.; Hirata, K.; Yamada, M.; Hamamori, Y.; Matsuda, Y.; Akita, H.; Yokoyama, M. Lysophosphatidylcholine inhibits bradykinin-induced phosphoinositide hydrolysis and calcium transients in cultured bovine aortic endothelial cells. Circ. Res. 1992, 71, 1410–1421. [Google Scholar] [CrossRef]
- Miwa, Y.; Hirata, K.; Kawashima, S.; Akita, H.; Yokoyama, M. Lysophosphatidylcholine inhibits receptor-mediated Ca2+ mobilization in intact endothelial cells of rabbit aorta. Arter. Thromb. Vasc. Biol. 1997, 17, 1561–1567. [Google Scholar] [CrossRef]
- Feron, O.; Dessy, C.; Moniotte, S.; Desager, J.P.; Balligand, J.L. Hypercholesterolemia decreases nitric oxide production by promoting the interaction of caveolin and endothelial nitric oxide synthase. J. Clin. Investig. 1999, 103, 897–905. [Google Scholar] [CrossRef]
- Nielsen, L.B. Transfer of low density lipoprotein into the arterial wall and risk of atherosclerosis. Atherosclerosis 1996, 123, 1–15. [Google Scholar] [CrossRef]
- Bolanle, I.O.; de Liedekerke Beaufort, G.C.; Weinberg, P.D. Transcytosis of LDL Across Arterial Endothelium: Mechanisms and Therapeutic Targets. Arter. Thromb. Vasc. Biol. 2025, 45, 468–480. [Google Scholar] [CrossRef]
- Boren, J.; Chapman, M.J.; Krauss, R.M.; Packard, C.J.; Bentzon, J.F.; Binder, C.J.; Daemen, M.J.; Demer, L.L.; Hegele, R.A.; Nicholls, S.J.; et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease: Pathophysiological, genetic, and therapeutic insights: A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2020, 41, 2313–2330. [Google Scholar] [CrossRef]
- Huang, L.; Chambliss, K.L.; Gao, X.; Yuhanna, I.S.; Behling-Kelly, E.; Bergaya, S.; Ahmed, M.; Michaely, P.; Luby-Phelps, K.; Darehshouri, A.; et al. SR-B1 drives endothelial cell LDL transcytosis via DOCK4 to promote atherosclerosis. Nature 2019, 569, 565–569. [Google Scholar] [CrossRef]
- Fung, K.Y.Y.; Ho, T.W.W.; Xu, Z.; Neculai, D.; Beauchemin, C.A.A.; Lee, W.L.; Fairn, G.D. Apolipoprotein A1 and high-density lipoprotein limit low-density lipoprotein transcytosis by binding SR-B1. J. Lipid Res. 2024, 65, 100530. [Google Scholar] [CrossRef] [PubMed]
- Nishi, K.; Itabe, H.; Uno, M.; Kitazato, K.T.; Horiguchi, H.; Shinno, K.; Nagahiro, S. Oxidized LDL in carotid plaques and plasma associates with plaque instability. Arter. Thromb. Vasc. Biol. 2002, 22, 1649–1654. [Google Scholar] [CrossRef]
- Ohlsson, B.G.; Englund, M.C.; Karlsson, A.L.; Knutsen, E.; Erixon, C.; Skribeck, H.; Liu, Y.; Bondjers, G.; Wiklund, O. Oxidized low density lipoprotein inhibits lipopolysaccharide-induced binding of nuclear factor-kappaB to DNA and the subsequent expression of tumor necrosis factor-alpha and interleukin-1beta in macrophages. J. Clin. Investig. 1996, 98, 78–89. [Google Scholar] [CrossRef]
- Li, D.; Mehta, J.L. Upregulation of endothelial receptor for oxidized LDL (LOX-1) by oxidized LDL and implications in apoptosis of human coronary artery endothelial cells: Evidence from use of antisense LOX-1 mRNA and chemical inhibitors. Arter. Thromb. Vasc. Biol. 2000, 20, 1116–1122. [Google Scholar] [CrossRef]
- Sawamura, T.; Kume, N.; Aoyama, T.; Moriwaki, H.; Hoshikawa, H.; Aiba, Y.; Tanaka, T.; Miwa, S.; Katsura, Y.; Kita, T.; et al. An endothelial receptor for oxidized low-density lipoprotein. Nature 1997, 386, 73–77. [Google Scholar] [CrossRef] [PubMed]
- Mehta, J.L.; Li, D.Y. Identification and autoregulation of receptor for OX-LDL in cultured human coronary artery endothelial cells. Biochem. Biophys. Res. Commun. 1998, 248, 511–514. [Google Scholar] [CrossRef] [PubMed]
- Cominacini, L.; Pasini, A.F.; Garbin, U.; Davoli, A.; Tosetti, M.L.; Campagnola, M.; Rigoni, A.; Pastorino, A.M.; Lo Cascio, V.; Sawamura, T. Oxidized low density lipoprotein (ox-LDL) binding to ox-LDL receptor-1 in endothelial cells induces the activation of NF-kappaB through an increased production of intracellular reactive oxygen species. J. Biol. Chem. 2000, 275, 12633–12638. [Google Scholar] [CrossRef]
- Galkina, E.; Ley, K. Vascular adhesion molecules in atherosclerosis. Arter. Thromb. Vasc. Biol. 2007, 27, 2292–2301. [Google Scholar] [CrossRef]
- Johnson-Tidey, R.R.; McGregor, J.L.; Taylor, P.R.; Poston, R.N. Increase in the adhesion molecule P-selectin in endothelium overlying atherosclerotic plaques. Coexpression with intercellular adhesion molecule-1. Am. J. Pathol. 1994, 144, 952–961. [Google Scholar] [PubMed]
- Collins, R.G.; Velji, R.; Guevara, N.V.; Hicks, M.J.; Chan, L.; Beaudet, A.L. P-Selectin or intercellular adhesion molecule (ICAM)-1 deficiency substantially protects against atherosclerosis in apolipoprotein E-deficient mice. J. Exp. Med. 2000, 191, 189–194. [Google Scholar] [CrossRef] [PubMed]
- Dong, Z.M.; Brown, A.A.; Wagner, D.D. Prominent role of P-selectin in the development of advanced atherosclerosis in ApoE-deficient mice. Circulation 2000, 101, 2290–2295. [Google Scholar] [CrossRef]
- Dong, Z.M.; Chapman, S.M.; Brown, A.A.; Frenette, P.S.; Hynes, R.O.; Wagner, D.D. The combined role of P- and E-selectins in atherosclerosis. J. Clin. Investig. 1998, 102, 145–152. [Google Scholar] [CrossRef]
- Davies, M.J.; Gordon, J.L.; Gearing, A.J.; Pigott, R.; Woolf, N.; Katz, D.; Kyriakopoulos, A. The expression of the adhesion molecules ICAM-1, VCAM-1, PECAM, and E-selectin in human atherosclerosis. J. Pathol. 1993, 171, 223–229. [Google Scholar] [CrossRef]
- Gerhardt, T.; Ley, K. Monocyte trafficking across the vessel wall. Cardiovasc. Res. 2015, 107, 321–330. [Google Scholar] [CrossRef]
- Cybulsky, M.I.; Gimbrone, M.A., Jr. Endothelial expression of a mononuclear leukocyte adhesion molecule during atherogenesis. Science 1991, 251, 788–791. [Google Scholar] [CrossRef]
- O’Brien, K.D.; Allen, M.D.; McDonald, T.O.; Chait, A.; Harlan, J.M.; Fishbein, D.; McCarty, J.; Ferguson, M.; Hudkins, K.; Benjamin, C.D.; et al. Vascular cell adhesion molecule-1 is expressed in human coronary atherosclerotic plaques. Implications for the mode of progression of advanced coronary atherosclerosis. J. Clin. Investig. 1993, 92, 945–951. [Google Scholar] [CrossRef] [PubMed]
- Nageh, M.F.; Sandberg, E.T.; Marotti, K.R.; Lin, A.H.; Melchior, E.P.; Bullard, D.C.; Beaudet, A.L. Deficiency of inflammatory cell adhesion molecules protects against atherosclerosis in mice. Arter. Thromb. Vasc. Biol. 1997, 17, 1517–1520. [Google Scholar] [CrossRef]
- Kitagawa, K.; Matsumoto, M.; Sasaki, T.; Hashimoto, H.; Kuwabara, K.; Ohtsuki, T.; Hori, M. Involvement of ICAM-1 in the progression of atherosclerosis in APOE-knockout mice. Atherosclerosis 2002, 160, 305–310. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Cybulsky, M.I.; Gimbrone, M.A., Jr.; Libby, P. An atherogenic diet rapidly induces VCAM-1, a cytokine-regulatable mononuclear leukocyte adhesion molecule, in rabbit aortic endothelium. Arter. Thromb. 1993, 13, 197–204. [Google Scholar] [CrossRef]
- Cybulsky, M.I.; Iiyama, K.; Li, H.; Zhu, S.; Chen, M.; Iiyama, M.; Davis, V.; Gutierrez-Ramos, J.C.; Connelly, P.W.; Milstone, D.S. A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. J. Clin. Investig. 2001, 107, 1255–1262. [Google Scholar] [CrossRef]
- Zarbock, A.; Ley, K.; McEver, R.P.; Hidalgo, A. Leukocyte ligands for endothelial selectins: Specialized glycoconjugates that mediate rolling and signaling under flow. Blood 2011, 118, 6743–6751. [Google Scholar] [CrossRef]
- Li, D.; Chen, H.; Romeo, F.; Sawamura, T.; Saldeen, T.; Mehta, J.L. Statins modulate oxidized low-density lipoprotein-mediated adhesion molecule expression in human coronary artery endothelial cells: Role of LOX-1. J. Pharmacol. Exp. Ther. 2002, 302, 601–605. [Google Scholar] [CrossRef]
- Tacke, F.; Alvarez, D.; Kaplan, T.J.; Jakubzick, C.; Spanbroek, R.; Llodra, J.; Garin, A.; Liu, J.; Mack, M.; van Rooijen, N.; et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J. Clin. Investig. 2007, 117, 185–194. [Google Scholar] [CrossRef]
- Barron, H.V.; Cannon, C.P.; Murphy, S.A.; Braunwald, E.; Gibson, C.M. Association between white blood cell count, epicardial blood flow, myocardial perfusion, and clinical outcomes in the setting of acute myocardial infarction: A thrombolysis in myocardial infarction 10 substudy. Circulation 2000, 102, 2329–2334. [Google Scholar] [CrossRef]
- Stewart, R.A.; White, H.D.; Kirby, A.C.; Heritier, S.R.; Simes, R.J.; Nestel, P.J.; West, M.J.; Colquhoun, D.M.; Tonkin, A.M.; Long-Term Intervention With Pravastatin in Ischemic Disease Study Investigator. White blood cell count predicts reduction in coronary heart disease mortality with pravastatin. Circulation 2005, 111, 1756–1762. [Google Scholar] [CrossRef] [PubMed]
- Tani, S.; Nagao, K.; Anazawa, T.; Kawamata, H.; Furuya, S.; Takahashi, H.; Iida, K.; Matsumoto, M.; Washio, T.; Kumabe, N.; et al. Association of leukocyte subtype counts with coronary atherosclerotic regression following pravastatin treatment. Am. J. Cardiol. 2009, 104, 464–469. [Google Scholar] [CrossRef] [PubMed]
- Ley, K.; Miller, Y.I.; Hedrick, C.C. Monocyte and macrophage dynamics during atherogenesis. Arter. Thromb. Vasc. Biol. 2011, 31, 1506–1516. [Google Scholar] [CrossRef]
- Hilgendorf, I.; Swirski, F.K.; Robbins, C.S. Monocyte fate in atherosclerosis. Arter. Thromb. Vasc. Biol. 2015, 35, 272–279. [Google Scholar] [CrossRef]
- Geissmann, F.; Jung, S.; Littman, D.R. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 2003, 19, 71–82. [Google Scholar] [CrossRef] [PubMed]
- Mildner, A.; Kim, K.W.; Yona, S. Unravelling monocyte functions: From the guardians of health to the regulators of disease. Discov. Immunol. 2024, 3, kyae014. [Google Scholar] [CrossRef] [PubMed]
- Swirski, F.K.; Libby, P.; Aikawa, E.; Alcaide, P.; Luscinskas, F.W.; Weissleder, R.; Pittet, M.J. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J. Clin. Investig. 2007, 117, 195–205. [Google Scholar] [CrossRef]
- Robbins, C.S.; Chudnovskiy, A.; Rauch, P.J.; Figueiredo, J.L.; Iwamoto, Y.; Gorbatov, R.; Etzrodt, M.; Weber, G.F.; Ueno, T.; van Rooijen, N.; et al. Extramedullary hematopoiesis generates Ly-6C(high) monocytes that infiltrate atherosclerotic lesions. Circulation 2012, 125, 364–374. [Google Scholar] [CrossRef]
- Emami, H.; Singh, P.; MacNabb, M.; Vucic, E.; Lavender, Z.; Rudd, J.H.; Fayad, Z.A.; Lehrer-Graiwer, J.; Korsgren, M.; Figueroa, A.L.; et al. Splenic metabolic activity predicts risk of future cardiovascular events: Demonstration of a cardiosplenic axis in humans. JACC Cardiovasc. Imaging 2015, 8, 121–130. [Google Scholar] [CrossRef] [PubMed]
- Gleissner, C.A.; Leitinger, N.; Ley, K. Effects of native and modified low-density lipoproteins on monocyte recruitment in atherosclerosis. Hypertension 2007, 50, 276–283. [Google Scholar] [CrossRef]
- Han, K.H.; Han, K.O.; Green, S.R.; Quehenberger, O. Expression of the monocyte chemoattractant protein-1 receptor CCR2 is increased in hypercholesterolemia. Differential effects of plasma lipoproteins on monocyte function. J. Lipid Res. 1999, 40, 1053–1063. [Google Scholar] [CrossRef]
- Han, K.H.; Tangirala, R.K.; Green, S.R.; Quehenberger, O. Chemokine receptor CCR2 expression and monocyte chemoattractant protein-1-mediated chemotaxis in human monocytes. A regulatory role for plasma LDL. Arter. Thromb. Vasc. Biol. 1998, 18, 1983–1991. [Google Scholar] [CrossRef]
- Cushing, S.D.; Berliner, J.A.; Valente, A.J.; Territo, M.C.; Navab, M.; Parhami, F.; Gerrity, R.; Schwartz, C.J.; Fogelman, A.M. Minimally modified low density lipoprotein induces monocyte chemotactic protein 1 in human endothelial cells and smooth muscle cells. Proc. Natl. Acad. Sci. USA 1990, 87, 5134–5138. [Google Scholar] [CrossRef]
- Lee, H.; Shi, W.; Tontonoz, P.; Wang, S.; Subbanagounder, G.; Hedrick, C.C.; Hama, S.; Borromeo, C.; Evans, R.M.; Berliner, J.A.; et al. Role for peroxisome proliferator-activated receptor alpha in oxidized phospholipid-induced synthesis of monocyte chemotactic protein-1 and interleukin-8 by endothelial cells. Circ. Res. 2000, 87, 516–521. [Google Scholar] [CrossRef] [PubMed]
- Suriyaphol, P.; Fenske, D.; Zahringer, U.; Han, S.R.; Bhakdi, S.; Husmann, M. Enzymatically modified nonoxidized low-density lipoprotein induces interleukin-8 in human endothelial cells: Role of free fatty acids. Circulation 2002, 106, 2581–2587. [Google Scholar] [CrossRef]
- Parhami, F.; Fang, Z.T.; Fogelman, A.M.; Andalibi, A.; Territo, M.C.; Berliner, J.A. Minimally modified low density lipoprotein-induced inflammatory responses in endothelial cells are mediated by cyclic adenosine monophosphate. J. Clin. Investig. 1993, 92, 471–478. [Google Scholar] [CrossRef]
- Gerszten, R.E.; Garcia-Zepeda, E.A.; Lim, Y.C.; Yoshida, M.; Ding, H.A.; Gimbrone, M.A., Jr.; Luster, A.D.; Luscinskas, F.W.; Rosenzweig, A. MCP-1 and IL-8 trigger firm adhesion of monocytes to vascular endothelium under flow conditions. Nature 1999, 398, 718–723. [Google Scholar] [CrossRef]
- Gu, L.; Okada, Y.; Clinton, S.K.; Gerard, C.; Sukhova, G.K.; Libby, P.; Rollins, B.J. Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient mice. Mol. Cell 1998, 2, 275–281. [Google Scholar] [CrossRef]
- Boring, L.; Gosling, J.; Cleary, M.; Charo, I.F. Decreased lesion formation in CCR2-/- mice reveals a role for chemokines in the initiation of atherosclerosis. Nature 1998, 394, 894–897. [Google Scholar] [CrossRef]
- Boisvert, W.A.; Santiago, R.; Curtiss, L.K.; Terkeltaub, R.A. A leukocyte homologue of the IL-8 receptor CXCR-2 mediates the accumulation of macrophages in atherosclerotic lesions of LDL receptor-deficient mice. J. Clin. Investig. 1998, 101, 353–363. [Google Scholar] [CrossRef]
- Moore, K.J.; Tabas, I. Macrophages in the pathogenesis of atherosclerosis. Cell 2011, 145, 341–355. [Google Scholar] [CrossRef] [PubMed]
- Smith, J.D.; Trogan, E.; Ginsberg, M.; Grigaux, C.; Tian, J.; Miyata, M. Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc. Natl. Acad. Sci. USA 1995, 92, 8264–8268. [Google Scholar] [CrossRef] [PubMed]
- Qiao, J.H.; Tripathi, J.; Mishra, N.K.; Cai, Y.; Tripathi, S.; Wang, X.P.; Imes, S.; Fishbein, M.C.; Clinton, S.K.; Libby, P.; et al. Role of macrophage colony-stimulating factor in atherosclerosis: Studies of osteopetrotic mice. Am. J. Pathol. 1997, 150, 1687–1699. [Google Scholar]
- Rajavashisth, T.B.; Andalibi, A.; Territo, M.C.; Berliner, J.A.; Navab, M.; Fogelman, A.M.; Lusis, A.J. Induction of endothelial cell expression of granulocyte and macrophage colony-stimulating factors by modified low-density lipoproteins. Nature 1990, 344, 254–257. [Google Scholar] [CrossRef]
- Sinha, S.K.; Miikeda, A.; Fouladian, Z.; Mehrabian, M.; Edillor, C.; Shih, D.; Zhou, Z.; Paul, M.K.; Charugundla, S.; Davis, R.C.; et al. Local M-CSF (Macrophage Colony-Stimulating Factor) Expression Regulates Macrophage Proliferation and Apoptosis in Atherosclerosis. Arter. Thromb. Vasc. Biol. 2021, 41, 220–233. [Google Scholar] [CrossRef]
- Bjorkegren, J.L.M.; Lusis, A.J. Atherosclerosis: Recent developments. Cell 2022, 185, 1630–1645. [Google Scholar] [CrossRef] [PubMed]
- Yoshida, H.; Quehenberger, O.; Kondratenko, N.; Green, S.; Steinberg, D. Minimally oxidized low-density lipoprotein increases expression of scavenger receptor A, CD36, and macrosialin in resident mouse peritoneal macrophages. Arter. Thromb. Vasc. Biol. 1998, 18, 794–802. [Google Scholar] [CrossRef] [PubMed]
- Avogaro, P.; Bon, G.B.; Cazzolato, G. Presence of a modified low density lipoprotein in humans. Arteriosclerosis 1988, 8, 79–87. [Google Scholar] [CrossRef]
- Boren, J.; Olin, K.; Lee, I.; Chait, A.; Wight, T.N.; Innerarity, T.L. Identification of the principal proteoglycan-binding site in LDL. A single-point mutation in apo-B100 severely affects proteoglycan interaction without affecting LDL receptor binding. J. Clin. Investig. 1998, 101, 2658–2664. [Google Scholar] [CrossRef] [PubMed]
- Kaplan, M.; Aviram, M. Retention of oxidized LDL by extracellular matrix proteoglycans leads to its uptake by macrophages: An alternative approach to study lipoproteins cellular uptake. Arter. Thromb. Vasc. Biol. 2001, 21, 386–393. [Google Scholar] [CrossRef]
- Stewart, C.R.; Stuart, L.M.; Wilkinson, K.; van Gils, J.M.; Deng, J.; Halle, A.; Rayner, K.J.; Boyer, L.; Zhong, R.; Frazier, W.A.; et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat. Immunol. 2010, 11, 155–161. [Google Scholar] [CrossRef]
- Sheedy, F.J.; Grebe, A.; Rayner, K.J.; Kalantari, P.; Ramkhelawon, B.; Carpenter, S.B.; Becker, C.E.; Ediriweera, H.N.; Mullick, A.E.; Golenbock, D.T.; et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 2013, 14, 812–820. [Google Scholar] [CrossRef]
- Kodama, T.; Reddy, P.; Kishimoto, C.; Krieger, M. Purification and characterization of a bovine acetyl low density lipoprotein receptor. Proc. Natl. Acad. Sci. USA 1988, 85, 9238–9242. [Google Scholar] [CrossRef]
- Kodama, T.; Freeman, M.; Rohrer, L.; Zabrecky, J.; Matsudaira, P.; Krieger, M. Type I macrophage scavenger receptor contains alpha-helical and collagen-like coiled coils. Nature 1990, 343, 531–535. [Google Scholar] [CrossRef]
- Seimon, T.A.; Nadolski, M.J.; Liao, X.; Magallon, J.; Nguyen, M.; Feric, N.T.; Koschinsky, M.L.; Harkewicz, R.; Witztum, J.L.; Tsimikas, S.; et al. Atherogenic lipids and lipoproteins trigger CD36-TLR2-dependent apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell Metab. 2010, 12, 467–482. [Google Scholar] [CrossRef]
- Tian, K.; Xu, Y.; Sahebkar, A.; Xu, S. CD36 in Atherosclerosis: Pathophysiological Mechanisms and Therapeutic Implications. Curr. Atheroscler. Rep. 2020, 22, 59. [Google Scholar] [CrossRef]
- Febbraio, M.; Hajjar, D.P.; Silverstein, R.L. CD36: A class B scavenger receptor involved in angiogenesis, atherosclerosis, inflammation, and lipid metabolism. J. Clin. Investig. 2001, 108, 785–791. [Google Scholar] [CrossRef] [PubMed]
- de Winther, M.P.; van Dijk, K.W.; Havekes, L.M.; Hofker, M.H. Macrophage scavenger receptor class A: A multifunctional receptor in atherosclerosis. Arter. Thromb. Vasc. Biol. 2000, 20, 290–297. [Google Scholar] [CrossRef] [PubMed]
- Schaeffer, D.F.; Riazy, M.; Parhar, K.S.; Chen, J.H.; Duronio, V.; Sawamura, T.; Steinbrecher, U.P. LOX-1 augments oxLDL uptake by lysoPC-stimulated murine macrophages but is not required for oxLDL clearance from plasma. J. Lipid Res. 2009, 50, 1676–1684. [Google Scholar] [CrossRef] [PubMed]
- Talle, M.A.; Rao, P.E.; Westberg, E.; Allegar, N.; Makowski, M.; Mittler, R.S.; Goldstein, G. Patterns of antigenic expression on human monocytes as defined by monoclonal antibodies. Cell Immunol. 1983, 78, 83–99. [Google Scholar] [CrossRef]
- Rahaman, S.O.; Lennon, D.J.; Febbraio, M.; Podrez, E.A.; Hazen, S.L.; Silverstein, R.L. A CD36-dependent signaling cascade is necessary for macrophage foam cell formation. Cell Metab. 2006, 4, 211–221. [Google Scholar] [CrossRef]
- Febbraio, M.; Podrez, E.A.; Smith, J.D.; Hajjar, D.P.; Hazen, S.L.; Hoff, H.F.; Sharma, K.; Silverstein, R.L. Targeted disruption of the class B scavenger receptor CD36 protects against atherosclerotic lesion development in mice. J. Clin. Investig. 2000, 105, 1049–1056. [Google Scholar] [CrossRef]
- Moore, K.J.; Kunjathoor, V.V.; Koehn, S.L.; Manning, J.J.; Tseng, A.A.; Silver, J.M.; McKee, M.; Freeman, M.W. Loss of receptor-mediated lipid uptake via scavenger receptor A or CD36 pathways does not ameliorate atherosclerosis in hyperlipidemic mice. J. Clin. Investig. 2005, 115, 2192–2201. [Google Scholar] [CrossRef]
- Manning-Tobin, J.J.; Moore, K.J.; Seimon, T.A.; Bell, S.A.; Sharuk, M.; Alvarez-Leite, J.I.; de Winther, M.P.; Tabas, I.; Freeman, M.W. Loss of SR-A and CD36 activity reduces atherosclerotic lesion complexity without abrogating foam cell formation in hyperlipidemic mice. Arter. Thromb. Vasc. Biol. 2009, 29, 19–26. [Google Scholar] [CrossRef]
- Singh, R.K.; Haka, A.S.; Asmal, A.; Barbosa-Lorenzi, V.C.; Grosheva, I.; Chin, H.F.; Xiong, Y.; Hla, T.; Maxfield, F.R. TLR4 (Toll-Like Receptor 4)-Dependent Signaling Drives Extracellular Catabolism of LDL (Low-Density Lipoprotein) Aggregates. Arter. Thromb. Vasc. Biol. 2020, 40, 86–102. [Google Scholar] [CrossRef] [PubMed]
- Lin, H.P.; Singla, B.; Ahn, W.; Ghoshal, P.; Blahove, M.; Cherian-Shaw, M.; Chen, A.; Haller, A.; Hui, D.Y.; Dong, K.; et al. Receptor-independent fluid-phase macropinocytosis promotes arterial foam cell formation and atherosclerosis. Sci. Transl. Med. 2022, 14, eadd2376. [Google Scholar] [CrossRef]
- Tabas, I. Cholesterol and phospholipid metabolism in macrophages. Biochim. Biophys. Acta 2000, 1529, 164–174. [Google Scholar] [CrossRef] [PubMed]
- Ouimet, M.; Franklin, V.; Mak, E.; Liao, X.; Tabas, I.; Marcel, Y.L. Autophagy regulates cholesterol efflux from macrophage foam cells via lysosomal acid lipase. Cell Metab. 2011, 13, 655–667. [Google Scholar] [CrossRef] [PubMed]
- Remaley, A.T.; Stonik, J.A.; Demosky, S.J.; Neufeld, E.B.; Bocharov, A.V.; Vishnyakova, T.G.; Eggerman, T.L.; Patterson, A.P.; Duverger, N.J.; Santamarina-Fojo, S.; et al. Apolipoprotein specificity for lipid efflux by the human ABCAI transporter. Biochem. Biophys. Res. Commun. 2001, 280, 818–823. [Google Scholar] [CrossRef]
- Kennedy, M.A.; Barrera, G.C.; Nakamura, K.; Baldan, A.; Tarr, P.; Fishbein, M.C.; Frank, J.; Francone, O.L.; Edwards, P.A. ABCG1 has a critical role in mediating cholesterol efflux to HDL and preventing cellular lipid accumulation. Cell Metab. 2005, 1, 121–131. [Google Scholar] [CrossRef]
- Brown, A.J.; Jessup, W. Oxysterols and atherosclerosis. Atherosclerosis 1999, 142, 1–28. [Google Scholar] [CrossRef]
- Janowski, B.A.; Willy, P.J.; Devi, T.R.; Falck, J.R.; Mangelsdorf, D.J. An oxysterol signalling pathway mediated by the nuclear receptor LXR alpha. Nature 1996, 383, 728–731. [Google Scholar] [CrossRef]
- Liu, Y.; Wei, Z.; Ma, X.; Yang, X.; Chen, Y.; Sun, L.; Ma, C.; Miao, Q.R.; Hajjar, D.P.; Han, J.; et al. 25-Hydroxycholesterol activates the expression of cholesterol 25-hydroxylase in an LXR-dependent mechanism. J. Lipid Res. 2018, 59, 439–451. [Google Scholar] [CrossRef]
- Lehmann, J.M.; Kliewer, S.A.; Moore, L.B.; Smith-Oliver, T.A.; Oliver, B.B.; Su, J.L.; Sundseth, S.S.; Winegar, D.A.; Blanchard, D.E.; Spencer, T.A.; et al. Activation of the nuclear receptor LXR by oxysterols defines a new hormone response pathway. J. Biol. Chem. 1997, 272, 3137–3140. [Google Scholar] [CrossRef]
- Rasheed, A.; Cummins, C.L. Beyond the Foam Cell: The Role of LXRs in Preventing Atherogenesis. Int. J. Mol. Sci. 2018, 19, 2307. [Google Scholar] [CrossRef] [PubMed]
- Joseph, S.B.; Castrillo, A.; Laffitte, B.A.; Mangelsdorf, D.J.; Tontonoz, P. Reciprocal regulation of inflammation and lipid metabolism by liver X receptors. Nat. Med. 2003, 9, 213–219. [Google Scholar] [CrossRef]
- Tangirala, R.K.; Bischoff, E.D.; Joseph, S.B.; Wagner, B.L.; Walczak, R.; Laffitte, B.A.; Daige, C.L.; Thomas, D.; Heyman, R.A.; Mangelsdorf, D.J.; et al. Identification of macrophage liver X receptors as inhibitors of atherosclerosis. Proc. Natl. Acad. Sci. USA 2002, 99, 11896–11901. [Google Scholar] [CrossRef]
- Alshaikhli, A.; Vaqar, S. Tangier Disease. In StatPearls; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
- Oram, J.F. Tangier disease and ABCA1. Biochim. Biophys. Acta 2000, 1529, 321–330. [Google Scholar] [CrossRef]
- Ghisletti, S.; Huang, W.; Jepsen, K.; Benner, C.; Hardiman, G.; Rosenfeld, M.G.; Glass, C.K. Cooperative NCoR/SMRT interactions establish a corepressor-based strategy for integration of inflammatory and anti-inflammatory signaling pathways. Genes. Dev. 2009, 23, 681–693. [Google Scholar] [CrossRef] [PubMed]
- Thomas, D.G.; Doran, A.C.; Fotakis, P.; Westerterp, M.; Antonson, P.; Jiang, H.; Jiang, X.C.; Gustafsson, J.A.; Tabas, I.; Tall, A.R. LXR Suppresses Inflammatory Gene Expression and Neutrophil Migration through cis-Repression and Cholesterol Efflux. Cell Rep. 2018, 25, 3774–3785.e4. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; McDonald, J.G.; Aryal, B.; Canfran-Duque, A.; Goldberg, E.L.; Araldi, E.; Ding, W.; Fan, Y.; Thompson, B.M.; Singh, A.K.; et al. Desmosterol suppresses macrophage inflammasome activation and protects against vascular inflammation and atherosclerosis. Proc. Natl. Acad. Sci. USA 2021, 118, e2107682118. [Google Scholar] [CrossRef]
- Spann, N.J.; Garmire, L.X.; McDonald, J.G.; Myers, D.S.; Milne, S.B.; Shibata, N.; Reichart, D.; Fox, J.N.; Shaked, I.; Heudobler, D.; et al. Regulated accumulation of desmosterol integrates macrophage lipid metabolism and inflammatory responses. Cell 2012, 151, 138–152. [Google Scholar] [CrossRef]
- Allahverdian, S.; Chehroudi, A.C.; McManus, B.M.; Abraham, T.; Francis, G.A. Contribution of intimal smooth muscle cells to cholesterol accumulation and macrophage-like cells in human atherosclerosis. Circulation 2014, 129, 1551–1559. [Google Scholar] [CrossRef]
- Feil, S.; Fehrenbacher, B.; Lukowski, R.; Essmann, F.; Schulze-Osthoff, K.; Schaller, M.; Feil, R. Transdifferentiation of vascular smooth muscle cells to macrophage-like cells during atherogenesis. Circ. Res. 2014, 115, 662–667. [Google Scholar] [CrossRef]
- Shankman, L.S.; Gomez, D.; Cherepanova, O.A.; Salmon, M.; Alencar, G.F.; Haskins, R.M.; Swiatlowska, P.; Newman, A.A.; Greene, E.S.; Straub, A.C.; et al. KLF4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat. Med. 2015, 21, 628–637. [Google Scholar] [CrossRef]
- Wang, Y.; Dubland, J.A.; Allahverdian, S.; Asonye, E.; Sahin, B.; Jaw, J.E.; Sin, D.D.; Seidman, M.A.; Leeper, N.J.; Francis, G.A. Smooth Muscle Cells Contribute the Majority of Foam Cells in ApoE (Apolipoprotein E)-Deficient Mouse Atherosclerosis. Arter. Thromb. Vasc. Biol. 2019, 39, 876–887. [Google Scholar] [CrossRef]
- Dubland, J.A.; Allahverdian, S.; Besler, K.J.; Ortega, C.; Wang, Y.; Pryma, C.S.; Boukais, K.; Chan, T.; Seidman, M.A.; Francis, G.A. Low LAL (Lysosomal Acid Lipase) Expression by Smooth Muscle Cells Relative to Macrophages as a Mechanism for Arterial Foam Cell Formation. Arter. Thromb. Vasc. Biol. 2021, 41, e354–e368. [Google Scholar] [CrossRef] [PubMed]
- Robichaud, S.; Rasheed, A.; Pietrangelo, A.; Doyoung Kim, A.; Boucher, D.M.; Emerton, C.; Vijithakumar, V.; Gharibeh, L.; Fairman, G.; Mak, E.; et al. Autophagy Is Differentially Regulated in Leukocyte and Nonleukocyte Foam Cells During Atherosclerosis. Circ. Res. 2022, 130, 831–847. [Google Scholar] [CrossRef] [PubMed]
- Hotamisligil, G.S. Endoplasmic reticulum stress and atherosclerosis. Nat. Med. 2010, 16, 396–399. [Google Scholar] [CrossRef]
- Feng, B.; Yao, P.M.; Li, Y.; Devlin, C.M.; Zhang, D.; Harding, H.P.; Sweeney, M.; Rong, J.X.; Kuriakose, G.; Fisher, E.A.; et al. The endoplasmic reticulum is the site of cholesterol-induced cytotoxicity in macrophages. Nat. Cell Biol. 2003, 5, 781–792. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Navia-Pelaez, J.M.; Choi, S.H.; Miller, Y.I. Macrophage inflammarafts in atherosclerosis. Curr. Opin. Lipidol. 2023, 34, 189–195. [Google Scholar] [CrossRef]
- Navia-Pelaez, J.M.; Agatisa-Boyle, C.; Choi, S.H.; Sak Kim, Y.; Li, S.; Alekseeva, E.; Weldy, K.; Miller, Y.I. Differential Expression of Inflammarafts in Macrophage Foam Cells and in Nonfoamy Macrophages in Atherosclerotic Lesions-Brief Report. Arter. Thromb. Vasc. Biol. 2023, 43, 323–329. [Google Scholar] [CrossRef]
- Canfran-Duque, A.; Rotllan, N.; Zhang, X.; Andres-Blasco, I.; Thompson, B.M.; Sun, J.; Price, N.L.; Fernandez-Fuertes, M.; Fowler, J.W.; Gomez-Coronado, D.; et al. Macrophage-Derived 25-Hydroxycholesterol Promotes Vascular Inflammation, Atherogenesis, and Lesion Remodeling. Circulation 2023, 147, 388–408. [Google Scholar] [CrossRef]
- Khan, T.J.; Semenkovich, C.F.; Zayed, M.A. De novo lipid synthesis in cardiovascular tissue and disease. Atherosclerosis 2025, 400, 119066. [Google Scholar] [CrossRef]
- Vogel, A.; Brunner, J.S.; Hajto, A.; Sharif, O.; Schabbauer, G. Lipid scavenging macrophages and inflammation. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2022, 1867, 159066. [Google Scholar] [CrossRef]
- Rasheed, A.; Rayner, K.J. Macrophage Responses to Environmental Stimuli During Homeostasis and Disease. Endocr. Rev. 2021, 42, 407–435. [Google Scholar] [CrossRef] [PubMed]
- Barrett, T.J. Macrophages in Atherosclerosis Regression. Arter. Thromb. Vasc. Biol. 2020, 40, 20–33. [Google Scholar] [CrossRef]
- Nomura, M.; Liu, J.; Yu, Z.X.; Yamazaki, T.; Yan, Y.; Kawagishi, H.; Rovira, II; Liu, C.; Wolfgang, M.J.; Mukouyama, Y.S.; et al. Macrophage fatty acid oxidation inhibits atherosclerosis progression. J. Mol. Cell Cardiol. 2019, 127, 270–276. [Google Scholar] [CrossRef]
- Kanter, J.E.; Kramer, F.; Barnhart, S.; Averill, M.M.; Vivekanandan-Giri, A.; Vickery, T.; Li, L.O.; Becker, L.; Yuan, W.; Chait, A.; et al. Diabetes promotes an inflammatory macrophage phenotype and atherosclerosis through acyl-CoA synthetase 1. Proc. Natl. Acad. Sci. USA 2012, 109, E715–E724. [Google Scholar] [CrossRef] [PubMed]
- Wen, H.; Gris, D.; Lei, Y.; Jha, S.; Zhang, L.; Huang, M.T.; Brickey, W.J.; Ting, J.P. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat. Immunol. 2011, 12, 408–415. [Google Scholar] [CrossRef] [PubMed]
- Thorne, R.F.; Ralston, K.J.; de Bock, C.E.; Mhaidat, N.M.; Zhang, X.D.; Boyd, A.W.; Burns, G.F. Palmitoylation of CD36/FAT regulates the rate of its post-transcriptional processing in the endoplasmic reticulum. Biochim. Biophys. Acta 2010, 1803, 1298–1307. [Google Scholar] [CrossRef]
- Kumano-Kuramochi, M.; Xie, Q.; Kajiwara, S.; Komba, S.; Minowa, T.; Machida, S. Lectin-like oxidized LDL receptor-1 is palmitoylated and internalizes ligands via caveolae/raft-dependent endocytosis. Biochem. Biophys. Res. Commun. 2013, 434, 594–599. [Google Scholar] [CrossRef]
- Dessi, M.; Noce, A.; Bertucci, P.; Manca di Villahermosa, S.; Zenobi, R.; Castagnola, V.; Addessi, E.; Di Daniele, N. Atherosclerosis, dyslipidemia, and inflammation: The significant role of polyunsaturated Fatty acids. ISRN Inflamm. 2013, 2013, 191823. [Google Scholar] [CrossRef]
- Wang, B.; Wu, L.; Chen, J.; Dong, L.; Chen, C.; Wen, Z.; Hu, J.; Fleming, I.; Wang, D.W. Metabolism pathways of arachidonic acids: Mechanisms and potential therapeutic targets. Signal Transduct. Target. Ther. 2021, 6, 94. [Google Scholar] [CrossRef]
- Yokode, M.; Kita, T.; Kikawa, Y.; Ogorochi, T.; Narumiya, S.; Kawai, C. Stimulated arachidonate metabolism during foam cell transformation of mouse peritoneal macrophages with oxidized low density lipoprotein. J. Clin. Investig. 1988, 81, 720–729. [Google Scholar] [CrossRef]
- Zhu, L.; Zhang, Y.; Guo, Z.; Wang, M. Cardiovascular Biology of Prostanoids and Drug Discovery. Arter. Thromb. Vasc. Biol. 2020, 40, 1454–1463. [Google Scholar] [CrossRef]
- Shahidi, F.; Ambigaipalan, P. Omega-3 Polyunsaturated Fatty Acids and Their Health Benefits. Annu. Rev. Food Sci. Technol. 2018, 9, 345–381. [Google Scholar] [CrossRef] [PubMed]
- Bang, H.O.; Dyerberg, J.; Sinclair, H.M. The composition of the Eskimo food in north western Greenland. Am. J. Clin. Nutr. 1980, 33, 2657–2661. [Google Scholar] [CrossRef] [PubMed]
- Davidson, M.; Bulkow, L.R.; Gellin, B.G. Cardiac mortality in Alaska’s indigenous and non-Native residents. Int. J. Epidemiol. 1993, 22, 62–71. [Google Scholar] [CrossRef] [PubMed]
- Albert, C.M.; Hennekens, C.H.; O’Donnell, C.J.; Ajani, U.A.; Carey, V.J.; Willett, W.C.; Ruskin, J.N.; Manson, J.E. Fish consumption and risk of sudden cardiac death. JAMA 1998, 279, 23–28. [Google Scholar] [CrossRef]
- Yokoyama, M.; Origasa, H.; Matsuzaki, M.; Matsuzawa, Y.; Saito, Y.; Ishikawa, Y.; Oikawa, S.; Sasaki, J.; Hishida, H.; Itakura, H.; et al. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): A randomised open-label, blinded endpoint analysis. Lancet 2007, 369, 1090–1098. [Google Scholar] [CrossRef]
- Bhatt, D.L.; Steg, P.G.; Miller, M.; Brinton, E.A.; Jacobson, T.A.; Ketchum, S.B.; Doyle, R.T., Jr.; Juliano, R.A.; Jiao, L.; Granowitz, C.; et al. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N. Engl. J. Med. 2019, 380, 11–22. [Google Scholar] [CrossRef]
- Basil, M.C.; Levy, B.D. Specialized pro-resolving mediators: Endogenous regulators of infection and inflammation. Nat. Rev. Immunol. 2016, 16, 51–67. [Google Scholar] [CrossRef]
- Stotts, C.; Corrales-Medina, V.F.; Rayner, K.J. Pneumonia-Induced Inflammation, Resolution and Cardiovascular Disease: Causes, Consequences and Clinical Opportunities. Circ. Res. 2023, 132, 751–774. [Google Scholar] [CrossRef]
- Ohira, T.; Arita, M.; Omori, K.; Recchiuti, A.; Van Dyke, T.E.; Serhan, C.N. Resolvin E1 receptor activation signals phosphorylation and phagocytosis. J. Biol. Chem. 2010, 285, 3451–3461. [Google Scholar] [CrossRef]
- Cash, J.L.; Hart, R.; Russ, A.; Dixon, J.P.; Colledge, W.H.; Doran, J.; Hendrick, A.G.; Carlton, M.B.; Greaves, D.R. Synthetic chemerin-derived peptides suppress inflammation through ChemR23. J. Exp. Med. 2008, 205, 767–775. [Google Scholar] [CrossRef]
- Rymut, N.; Heinz, J.; Sadhu, S.; Hosseini, Z.; Riley, C.O.; Marinello, M.; Maloney, J.; MacNamara, K.C.; Spite, M.; Fredman, G. Resolvin D1 promotes efferocytosis in aging by limiting senescent cell-induced MerTK cleavage. FASEB J. 2020, 34, 597–609. [Google Scholar] [CrossRef]
- Hosseini, Z.; Marinello, M.; Decker, C.; Sansbury, B.E.; Sadhu, S.; Gerlach, B.D.; Bossardi Ramos, R.; Adam, A.P.; Spite, M.; Fredman, G. Resolvin D1 Enhances Necroptotic Cell Clearance Through Promoting Macrophage Fatty Acid Oxidation and Oxidative Phosphorylation. Arter. Thromb. Vasc. Biol. 2021, 41, 1062–1075. [Google Scholar] [CrossRef]
- Arnardottir, H.; Thul, S.; Pawelzik, S.C.; Karadimou, G.; Artiach, G.; Gallina, A.L.; Mysdotter, V.; Carracedo, M.; Tarnawski, L.; Caravaca, A.S.; et al. The resolvin D1 receptor GPR32 transduces inflammation resolution and atheroprotection. J. Clin. Investig. 2021, 131, e142883. [Google Scholar] [CrossRef] [PubMed]
- Nordestgaard, B.G. Triglyceride-Rich Lipoproteins and Atherosclerotic Cardiovascular Disease: New Insights From Epidemiology, Genetics, and Biology. Circ. Res. 2016, 118, 547–563. [Google Scholar] [CrossRef] [PubMed]
- Bjornson, E.; Adiels, M.; Gummesson, A.; Taskinen, M.R.; Burgess, S.; Packard, C.J.; Boren, J. Quantifying Triglyceride-Rich Lipoprotein Atherogenicity, Associations With Inflammation, and Implications for Risk Assessment Using Non-HDL Cholesterol. J. Am. Coll. Cardiol. 2024, 84, 1328–1338. [Google Scholar] [CrossRef]
- Tybjaerg-Hansen, A.; Nordestgaard, B.G.; Christoffersen, M. Triglyceride-rich remnant lipoproteins are more atherogenic than LDL per particle: Is this important? Eur. Heart J. 2023, 44, 4196–4198. [Google Scholar] [CrossRef]
- Taskinen, M.R.; Boren, J. Why Is Apolipoprotein CIII Emerging as a Novel Therapeutic Target to Reduce the Burden of Cardiovascular Disease? Curr. Atheroscler. Rep. 2016, 18, 59. [Google Scholar] [CrossRef] [PubMed]
- Bornfeldt, K.E. Apolipoprotein C3: Form begets function. J. Lipid Res. 2024, 65, 100475. [Google Scholar] [CrossRef]
- Kanter, J.E.; Shao, B.; Kramer, F.; Barnhart, S.; Shimizu-Albergine, M.; Vaisar, T.; Graham, M.J.; Crooke, R.M.; Manuel, C.R.; Haeusler, R.A.; et al. Increased apolipoprotein C3 drives cardiovascular risk in type 1 diabetes. J. Clin. Investig. 2019, 129, 4165–4179. [Google Scholar] [CrossRef]
- Qamar, A.; Khetarpal, S.A.; Khera, A.V.; Qasim, A.; Rader, D.J.; Reilly, M.P. Plasma apolipoprotein C-III levels, triglycerides, and coronary artery calcification in type 2 diabetics. Arter. Thromb. Vasc. Biol. 2015, 35, 1880–1888. [Google Scholar] [CrossRef] [PubMed]
- Pollin, T.I.; Damcott, C.M.; Shen, H.; Ott, S.H.; Shelton, J.; Horenstein, R.B.; Post, W.; McLenithan, J.C.; Bielak, L.F.; Peyser, P.A.; et al. A null mutation in human APOC3 confers a favorable plasma lipid profile and apparent cardioprotection. Science 2008, 322, 1702–1705. [Google Scholar] [CrossRef] [PubMed]
- Wulff, A.B.; Nordestgaard, B.G.; Tybjaerg-Hansen, A. APOC3 Loss-of-Function Mutations, Remnant Cholesterol, Low-Density Lipoprotein Cholesterol, and Cardiovascular Risk: Mediation- and Meta-Analyses of 137 895 Individuals. Arter. Thromb. Vasc. Biol. 2018, 38, 660–668. [Google Scholar] [CrossRef]
- Jorgensen, A.B.; Frikke-Schmidt, R.; Nordestgaard, B.G.; Tybjaerg-Hansen, A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N. Engl. J. Med. 2014, 371, 32–41. [Google Scholar] [CrossRef] [PubMed]
- Sundaram, M.; Zhong, S.; Bou Khalil, M.; Links, P.H.; Zhao, Y.; Iqbal, J.; Hussain, M.M.; Parks, R.J.; Wang, Y.; Yao, Z. Expression of apolipoprotein C-III in McA-RH7777 cells enhances VLDL assembly and secretion under lipid-rich conditions. J. Lipid Res. 2010, 51, 150–161. [Google Scholar] [CrossRef]
- Brown, W.V.; Baginsky, M.L. Inhibition of lipoprotein lipase by an apoprotein of human very low density lipoprotein. Biochem. Biophys. Res. Commun. 1972, 46, 375–382. [Google Scholar] [CrossRef]
- Windler, E.; Havel, R.J. Inhibitory effects of C apolipoproteins from rats and humans on the uptake of triglyceride-rich lipoproteins and their remnants by the perfused rat liver. J. Lipid Res. 1985, 26, 556–565. [Google Scholar] [CrossRef]
- Mendivil, C.O.; Rimm, E.B.; Furtado, J.; Chiuve, S.E.; Sacks, F.M. Low-density lipoproteins containing apolipoprotein C-III and the risk of coronary heart disease. Circulation 2011, 124, 2065–2072. [Google Scholar] [CrossRef]
- Emerging Risk Factors, C.; Erqou, S.; Kaptoge, S.; Perry, P.L.; Di Angelantonio, E.; Thompson, A.; White, I.R.; Marcovina, S.M.; Collins, R.; Thompson, S.G.; et al. Lipoprotein(a) concentration and the risk of coronary heart disease, stroke, and nonvascular mortality. JAMA 2009, 302, 412–423. [Google Scholar] [CrossRef]
- Bellomo, T.R.; Bramel, E.E.; Lee, J.; Urbut, S.; Flores, A.; Yu, Z.; Koyama, S.; Truong, B.; Haidermota, S.; Eagleton, M.J.; et al. Evaluation of Lipoprotein(a) as a Prognostic Marker of Extracoronary Atherosclerotic Vascular Disease Progression. Circulation 2025, 152, 585–598. [Google Scholar] [CrossRef]
- Marcovina, S.M.; Koschinsky, M.L. Lipoprotein(a) as a risk factor for coronary artery disease. Am. J. Cardiol. 1998, 82, 57U–66U, discussion 86U. [Google Scholar] [CrossRef] [PubMed]
- Reyes-Soffer, G.; Ginsberg, H.N.; Berglund, L.; Duell, P.B.; Heffron, S.P.; Kamstrup, P.R.; Lloyd-Jones, D.M.; Marcovina, S.M.; Yeang, C.; Koschinsky, M.L.; et al. Lipoprotein(a): A Genetically Determined, Causal, and Prevalent Risk Factor for Atherosclerotic Cardiovascular Disease: A Scientific Statement From the American Heart Association. Arter. Thromb. Vasc. Biol. 2022, 42, e48–e60. [Google Scholar] [CrossRef]
- Bjornson, E.; Adiels, M.; Taskinen, M.R.; Burgess, S.; Chapman, M.J.; Packard, C.J.; Boren, J. Lipoprotein(a) Is Markedly More Atherogenic Than LDL: An Apolipoprotein B-Based Genetic Analysis. J. Am. Coll. Cardiol. 2024, 83, 385–395. [Google Scholar] [CrossRef]
- Tsimikas, S.; Bittner, V. Particle Number and Characteristics of Lipoprotein(a), LDL, and apoB: Perspectives on Contributions to ASCVD. J. Am. Coll. Cardiol. 2024, 83, 396–400. [Google Scholar] [CrossRef] [PubMed]
- Bergmark, C.; Dewan, A.; Orsoni, A.; Merki, E.; Miller, E.R.; Shin, M.J.; Binder, C.J.; Horkko, S.; Krauss, R.M.; Chapman, M.J.; et al. A novel function of lipoprotein [a] as a preferential carrier of oxidized phospholipids in human plasma. J. Lipid Res. 2008, 49, 2230–2239. [Google Scholar] [CrossRef]
- Tsimikas, S.; Brilakis, E.S.; Miller, E.R.; McConnell, J.P.; Lennon, R.J.; Kornman, K.S.; Witztum, J.L.; Berger, P.B. Oxidized phospholipids, Lp(a) lipoprotein, and coronary artery disease. N. Engl. J. Med. 2005, 353, 46–57. [Google Scholar] [CrossRef] [PubMed]
- Sotiriou, S.N.; Orlova, V.V.; Al-Fakhri, N.; Ihanus, E.; Economopoulou, M.; Isermann, B.; Bdeir, K.; Nawroth, P.P.; Preissner, K.T.; Gahmberg, C.G.; et al. Lipoprotein(a) in atherosclerotic plaques recruits inflammatory cells through interaction with Mac-1 integrin. FASEB J. 2006, 20, 559–561. [Google Scholar] [CrossRef]
- Schnitzler, J.G.; Hoogeveen, R.M.; Ali, L.; Prange, K.H.M.; Waissi, F.; van Weeghel, M.; Bachmann, J.C.; Versloot, M.; Borrelli, M.J.; Yeang, C.; et al. Atherogenic Lipoprotein(a) Increases Vascular Glycolysis, Thereby Facilitating Inflammation and Leukocyte Extravasation. Circ. Res. 2020, 126, 1346–1359. [Google Scholar] [CrossRef]
- Keesler, G.A.; Li, Y.; Skiba, P.J.; Fless, G.M.; Tabas, I. Macrophage foam cell lipoprotein(a)/apoprotein(a) receptor. Cell-surface localization, dependence of induction on new protein synthesis, and ligand specificity. Arter. Thromb. 1994, 14, 1337–1345. [Google Scholar] [CrossRef] [PubMed]
- Chaurasia, B.; Summers, S.A. Ceramides in Metabolism: Key Lipotoxic Players. Annu. Rev. Physiol. 2021, 83, 303–330. [Google Scholar] [CrossRef]
- Zietzer, A.; Dusing, P.; Reese, L.; Nickenig, G.; Jansen, F. Ceramide Metabolism in Cardiovascular Disease: A Network With High Therapeutic Potential. Arter. Thromb. Vasc. Biol. 2022, 42, 1220–1228. [Google Scholar] [CrossRef] [PubMed]
- Summers, S.A. Could Ceramides Become the New Cholesterol? Cell Metab. 2018, 27, 276–280. [Google Scholar] [CrossRef]
- Havulinna, A.S.; Sysi-Aho, M.; Hilvo, M.; Kauhanen, D.; Hurme, R.; Ekroos, K.; Salomaa, V.; Laaksonen, R. Circulating Ceramides Predict Cardiovascular Outcomes in the Population-Based FINRISK 2002 Cohort. Arter. Thromb. Vasc. Biol. 2016, 36, 2424–2430. [Google Scholar] [CrossRef]
- Meikle, P.J.; Wong, G.; Tsorotes, D.; Barlow, C.K.; Weir, J.M.; Christopher, M.J.; MacIntosh, G.L.; Goudey, B.; Stern, L.; Kowalczyk, A.; et al. Plasma lipidomic analysis of stable and unstable coronary artery disease. Arter. Thromb. Vasc. Biol. 2011, 31, 2723–2732. [Google Scholar] [CrossRef]
- Laaksonen, R.; Ekroos, K.; Sysi-Aho, M.; Hilvo, M.; Vihervaara, T.; Kauhanen, D.; Suoniemi, M.; Hurme, R.; Marz, W.; Scharnagl, H.; et al. Plasma ceramides predict cardiovascular death in patients with stable coronary artery disease and acute coronary syndromes beyond LDL-cholesterol. Eur. Heart J. 2016, 37, 1967–1976. [Google Scholar] [CrossRef]
- Anroedh, S.; Hilvo, M.; Akkerhuis, K.M.; Kauhanen, D.; Koistinen, K.; Oemrawsingh, R.; Serruys, P.; van Geuns, R.J.; Boersma, E.; Laaksonen, R.; et al. Plasma concentrations of molecular lipid species predict long-term clinical outcome in coronary artery disease patients. J. Lipid Res. 2018, 59, 1729–1737. [Google Scholar] [CrossRef]
- Hilvo, M.; Meikle, P.J.; Pedersen, E.R.; Tell, G.S.; Dhar, I.; Brenner, H.; Schottker, B.; Laaperi, M.; Kauhanen, D.; Koistinen, K.M.; et al. Development and validation of a ceramide- and phospholipid-based cardiovascular risk estimation score for coronary artery disease patients. Eur. Heart J. 2020, 41, 371–380. [Google Scholar] [CrossRef] [PubMed]
- Gencer, B.; Morrow, D.A.; Braunwald, E.; Goodrich, E.L.; Hilvo, M.; Kauhanen, D.; Sabatine, M.S.; Laaksonen, R.; O’Donoghue, M.L. Plasma ceramide and phospholipid-based risk score and the risk of cardiovascular death in patients after acute coronary syndrome. Eur. J. Prev. Cardiol. 2022, 29, 895–902. [Google Scholar] [CrossRef] [PubMed]
- Schissel, S.L.; Tweedie-Hardman, J.; Rapp, J.H.; Graham, G.; Williams, K.J.; Tabas, I. Rabbit aorta and human atherosclerotic lesions hydrolyze the sphingomyelin of retained low-density lipoprotein. Proposed role for arterial-wall sphingomyelinase in subendothelial retention and aggregation of atherogenic lipoproteins. J. Clin. Investig. 1996, 98, 1455–1464. [Google Scholar] [CrossRef]
- Cheng, J.M.; Suoniemi, M.; Kardys, I.; Vihervaara, T.; de Boer, S.P.; Akkerhuis, K.M.; Sysi-Aho, M.; Ekroos, K.; Garcia-Garcia, H.M.; Oemrawsingh, R.M.; et al. Plasma concentrations of molecular lipid species in relation to coronary plaque characteristics and cardiovascular outcome: Results of the ATHEROREMO-IVUS study. Atherosclerosis 2015, 243, 560–566. [Google Scholar] [CrossRef]
- Li, W.; Yang, X.; Xing, S.; Bian, F.; Yao, W.; Bai, X.; Zheng, T.; Wu, G.; Jin, S. Endogenous ceramide contributes to the transcytosis of oxLDL across endothelial cells and promotes its subendothelial retention in vascular wall. Oxid. Med. Cell Longev. 2014, 2014, 823071. [Google Scholar] [CrossRef] [PubMed]
- Walters, M.J.; Wrenn, S.P. Effect of sphingomyelinase-mediated generation of ceramide on aggregation of low-density lipoprotein. Langmuir 2008, 24, 9642–9647. [Google Scholar] [CrossRef]
- Devlin, C.M.; Leventhal, A.R.; Kuriakose, G.; Schuchman, E.H.; Williams, K.J.; Tabas, I. Acid sphingomyelinase promotes lipoprotein retention within early atheromata and accelerates lesion progression. Arter. Thromb. Vasc. Biol. 2008, 28, 1723–1730. [Google Scholar] [CrossRef]
- Glaros, E.N.; Kim, W.S.; Quinn, C.M.; Jessup, W.; Rye, K.A.; Garner, B. Myriocin slows the progression of established atherosclerotic lesions in apolipoprotein E gene knockout mice. J. Lipid Res. 2008, 49, 324–331. [Google Scholar] [CrossRef]
- Gao, D.; Pararasa, C.; Dunston, C.R.; Bailey, C.J.; Griffiths, H.R. Palmitate promotes monocyte atherogenicity via de novo ceramide synthesis. Free Radic. Biol. Med. 2012, 53, 796–806. [Google Scholar] [CrossRef]
- Tsimikas, S.; Witztum, J.L. Oxidized phospholipids in cardiovascular disease. Nat. Rev. Cardiol. 2024, 21, 170–191. [Google Scholar] [CrossRef] [PubMed]
- Tsimikas, S.; Bergmark, C.; Beyer, R.W.; Patel, R.; Pattison, J.; Miller, E.; Juliano, J.; Witztum, J.L. Temporal increases in plasma markers of oxidized low-density lipoprotein strongly reflect the presence of acute coronary syndromes. J. Am. Coll. Cardiol. 2003, 41, 360–370. [Google Scholar] [CrossRef]
- Lee, S.; Birukov, K.G.; Romanoski, C.E.; Springstead, J.R.; Lusis, A.J.; Berliner, J.A. Role of phospholipid oxidation products in atherosclerosis. Circ. Res. 2012, 111, 778–799. [Google Scholar] [CrossRef] [PubMed]
- Que, X.; Hung, M.Y.; Yeang, C.; Gonen, A.; Prohaska, T.A.; Sun, X.; Diehl, C.; Maatta, A.; Gaddis, D.E.; Bowden, K.; et al. Oxidized phospholipids are proinflammatory and proatherogenic in hypercholesterolaemic mice. Nature 2018, 558, 301–306. [Google Scholar] [CrossRef]
- Boullier, A.; Gillotte, K.L.; Horkko, S.; Green, S.R.; Friedman, P.; Dennis, E.A.; Witztum, J.L.; Steinberg, D.; Quehenberger, O. The binding of oxidized low density lipoprotein to mouse CD36 is mediated in part by oxidized phospholipids that are associated with both the lipid and protein moieties of the lipoprotein. J. Biol. Chem. 2000, 275, 9163–9169. [Google Scholar] [CrossRef]
- Gillotte-Taylor, K.; Boullier, A.; Witztum, J.L.; Steinberg, D.; Quehenberger, O. Scavenger receptor class B type I as a receptor for oxidized low density lipoprotein. J. Lipid Res. 2001, 42, 1474–1482. [Google Scholar] [CrossRef]
- Kadl, A.; Meher, A.K.; Sharma, P.R.; Lee, M.Y.; Doran, A.C.; Johnstone, S.R.; Elliott, M.R.; Gruber, F.; Han, J.; Chen, W.; et al. Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ. Res. 2010, 107, 737–746. [Google Scholar] [CrossRef]
- Miller, Y.I.; Shyy, J.Y. Context-Dependent Role of Oxidized Lipids and Lipoproteins in Inflammation. Trends Endocrinol. Metab. 2017, 28, 143–152. [Google Scholar] [CrossRef]
- Ravandi, A.; Leibundgut, G.; Hung, M.Y.; Patel, M.; Hutchins, P.M.; Murphy, R.C.; Prasad, A.; Mahmud, E.; Miller, Y.I.; Dennis, E.A.; et al. Release and capture of bioactive oxidized phospholipids and oxidized cholesteryl esters during percutaneous coronary and peripheral arterial interventions in humans. J. Am. Coll. Cardiol. 2014, 63, 1961–1971. [Google Scholar] [CrossRef] [PubMed]
- Yan, F.X.; Li, H.M.; Li, S.X.; He, S.H.; Dai, W.P.; Li, Y.; Wang, T.T.; Shi, M.M.; Yuan, H.X.; Xu, Z.; et al. The oxidized phospholipid POVPC impairs endothelial function and vasodilation via uncoupling endothelial nitric oxide synthase. J. Mol. Cell Cardiol. 2017, 112, 40–48. [Google Scholar] [CrossRef]
- Lu, L.; Ye, Y.; Chen, Y.; Feng, L.; Huang, J.; Liang, Q.; Lan, Z.; Dong, Q.; Liu, X.; Li, Y.; et al. Oxidized phospholipid POVPC contributes to vascular calcification by triggering ferroptosis of vascular smooth muscle cells. FASEB J. 2024, 38, e23592. [Google Scholar] [CrossRef]
- Appleton, B.D.; Palmer, S.A.; Smith, H.P.; Stephens, L.E.; Major, A.S. Oxidized Phospholipid oxPAPC Alters Regulatory T-Cell Differentiation and Decreases Their Protective Function in Atherosclerosis in Mice. Arter. Thromb. Vasc. Biol. 2023, 43, 2119–2132. [Google Scholar] [CrossRef] [PubMed]
- Di Gioia, M.; Spreafico, R.; Springstead, J.R.; Mendelson, M.M.; Joehanes, R.; Levy, D.; Zanoni, I. Endogenous oxidized phospholipids reprogram cellular metabolism and boost hyperinflammation. Nat. Immunol. 2020, 21, 42–53. [Google Scholar] [CrossRef] [PubMed]
- van der Valk, F.M.; Bekkering, S.; Kroon, J.; Yeang, C.; Van den Bossche, J.; van Buul, J.D.; Ravandi, A.; Nederveen, A.J.; Verberne, H.J.; Scipione, C.; et al. Oxidized Phospholipids on Lipoprotein(a) Elicit Arterial Wall Inflammation and an Inflammatory Monocyte Response in Humans. Circulation 2016, 134, 611–624. [Google Scholar] [CrossRef]
- De Meyer, G.R.Y.; Zurek, M.; Puylaert, P.; Martinet, W. Programmed death of macrophages in atherosclerosis: Mechanisms and therapeutic targets. Nat. Rev. Cardiol. 2024, 21, 312–325. [Google Scholar] [CrossRef] [PubMed]
- Karunakaran, D.; Geoffrion, M.; Wei, L.; Gan, W.; Richards, L.; Shangari, P.; DeKemp, E.M.; Beanlands, R.A.; Perisic, L.; Maegdefessel, L.; et al. Targeting macrophage necroptosis for therapeutic and diagnostic interventions in atherosclerosis. Sci. Adv. 2016, 2, e1600224. [Google Scholar] [CrossRef]
- Karunakaran, D.; Nguyen, M.A.; Geoffrion, M.; Vreeken, D.; Lister, Z.; Cheng, H.S.; Otte, N.; Essebier, P.; Wyatt, H.; Kandiah, J.W.; et al. RIPK1 Expression Associates With Inflammation in Early Atherosclerosis in Humans and Can Be Therapeutically Silenced to Reduce NF-kappaB Activation and Atherogenesis in Mice. Circulation 2021, 143, 163–177. [Google Scholar] [CrossRef] [PubMed]
- Lin, J.; Li, H.; Yang, M.; Ren, J.; Huang, Z.; Han, F.; Huang, J.; Ma, J.; Zhang, D.; Zhang, Z.; et al. A role of RIP3-mediated macrophage necrosis in atherosclerosis development. Cell Rep. 2013, 3, 200–210. [Google Scholar] [CrossRef] [PubMed]
- Rasheed, A.; Robichaud, S.; Nguyen, M.A.; Geoffrion, M.; Wyatt, H.; Cottee, M.L.; Dennison, T.; Pietrangelo, A.; Lee, R.; Lagace, T.A.; et al. Loss of MLKL (Mixed Lineage Kinase Domain-Like Protein) Decreases Necrotic Core but Increases Macrophage Lipid Accumulation in Atherosclerosis. Arter. Thromb. Vasc. Biol. 2020, 40, 1155–1167. [Google Scholar] [CrossRef]
- Jiang, M.; Sun, X.; Liu, S.; Tang, Y.; Shi, Y.; Bai, Y.; Wang, Y.; Yang, Q.; Yang, Q.; Jiang, W.; et al. Caspase-11-Gasdermin D-Mediated Pyroptosis Is Involved in the Pathogenesis of Atherosclerosis. Front. Pharmacol. 2021, 12, 657486. [Google Scholar] [CrossRef]
- Shi, X.; Xie, W.L.; Kong, W.W.; Chen, D.; Qu, P. Expression of the NLRP3 Inflammasome in Carotid Atherosclerosis. J. Stroke Cerebrovasc. Dis. 2015, 24, 2455–2466. [Google Scholar] [CrossRef]
- Rajamaki, K.; Mayranpaa, M.I.; Risco, A.; Tuimala, J.; Nurmi, K.; Cuenda, A.; Eklund, K.K.; Oorni, K.; Kovanen, P.T. p38delta MAPK: A Novel Regulator of NLRP3 Inflammasome Activation With Increased Expression in Coronary Atherogenesis. Arter. Thromb. Vasc. Biol. 2016, 36, 1937–1946. [Google Scholar] [CrossRef]
- Zheng, F.; Xing, S.; Gong, Z.; Xing, Q. NLRP3 inflammasomes show high expression in aorta of patients with atherosclerosis. Heart Lung Circ. 2013, 22, 746–750. [Google Scholar] [CrossRef]
- Duewell, P.; Kono, H.; Rayner, K.J.; Sirois, C.M.; Vladimer, G.; Bauernfeind, F.G.; Abela, G.S.; Franchi, L.; Nunez, G.; Schnurr, M.; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464, 1357–1361. [Google Scholar] [CrossRef] [PubMed]
- Balasubramanian, A.; Hsu, A.Y.; Ghimire, L.; Tahir, M.; Devant, P.; Fontana, P.; Du, G.; Liu, X.; Fabin, D.; Kambara, H.; et al. The palmitoylation of gasdermin D directs its membrane translocation and pore formation during pyroptosis. Sci. Immunol. 2024, 9, eadn1452. [Google Scholar] [CrossRef]
- Du, G.; Healy, L.B.; David, L.; Walker, C.; El-Baba, T.J.; Lutomski, C.A.; Goh, B.; Gu, B.; Pi, X.; Devant, P.; et al. ROS-dependent S-palmitoylation activates cleaved and intact gasdermin D. Nature 2024, 630, 437–446. [Google Scholar] [CrossRef]
- Yalcinkaya, M.; Fotakis, P.; Liu, W.; Endo-Umeda, K.; Dou, H.; Abramowicz, S.; Xiao, T.; Libby, P.; Wang, N.; Tall, A.R.; et al. Cholesterol accumulation in macrophages drives NETosis in atherosclerotic plaques via IL-1beta secretion. Cardiovasc. Res. 2023, 119, 969–981. [Google Scholar] [CrossRef]
- Fuhrman, B.; Oiknine, J.; Aviram, M. Iron induces lipid peroxidation in cultured macrophages, increases their ability to oxidatively modify LDL, and affects their secretory properties. Atherosclerosis 1994, 111, 65–78. [Google Scholar] [CrossRef]
- Yuan, X.M.; Anders, W.L.; Olsson, A.G.; Brunk, U.T. Iron in human atheroma and LDL oxidation by macrophages following erythrophagocytosis. Atherosclerosis 1996, 124, 61–73. [Google Scholar] [CrossRef]
- Wang, L.J.; Lee, T.S.; Lee, F.Y.; Pai, R.C.; Chau, L.Y. Expression of heme oxygenase-1 in atherosclerotic lesions. Am. J. Pathol. 1998, 152, 711–720. [Google Scholar]
- Yuan, X.M.; Li, W.; Baird, S.K.; Carlsson, M.; Melefors, O. Secretion of ferritin by iron-laden macrophages and influence of lipoproteins. Free Radic. Res. 2004, 38, 1133–1142. [Google Scholar] [CrossRef]
- Bai, T.; Li, M.; Liu, Y.; Qiao, Z.; Wang, Z. Inhibition of ferroptosis alleviates atherosclerosis through attenuating lipid peroxidation and endothelial dysfunction in mouse aortic endothelial cell. Free Radic. Biol. Med. 2020, 160, 92–102. [Google Scholar] [CrossRef] [PubMed]
- Ward, N.C.; Watts, G.F.; Eckel, R.H. Statin Toxicity. Circ. Res. 2019, 124, 328–350. [Google Scholar] [CrossRef] [PubMed]
- Chou, R.; Cantor, A.; Dana, T.; Wagner, J.; Ahmed, A.Y.; Fu, R.; Ferencik, M. Statin Use for the Primary Prevention of Cardiovascular Disease in Adults: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 2022, 328, 754–771. [Google Scholar] [CrossRef] [PubMed]
- Force, U.S.P.S.T.; Mangione, C.M.; Barry, M.J.; Nicholson, W.K.; Cabana, M.; Chelmow, D.; Coker, T.R.; Davis, E.M.; Donahue, K.E.; Jaen, C.R.; et al. Statin Use for the Primary Prevention of Cardiovascular Disease in Adults: US Preventive Services Task Force Recommendation Statement. JAMA 2022, 328, 746–753. [Google Scholar] [CrossRef]
- Razavi, A.C.; Mehta, A.; Sperling, L.S. Statin therapy for the primary prevention of cardiovascular disease: Pros. Atherosclerosis 2022, 356, 41–45. [Google Scholar] [CrossRef]
- Fitchett, D.H.; Hegele, R.A.; Verma, S. Cardiology patient page. Statin intolerance. Circ. 2015, 131, e389–e391. [Google Scholar] [CrossRef]
- Bytyci, I.; Penson, P.E.; Mikhailidis, D.P.; Wong, N.D.; Hernandez, A.V.; Sahebkar, A.; Thompson, P.D.; Mazidi, M.; Rysz, J.; Pella, D.; et al. Prevalence of statin intolerance: A meta-analysis. Eur. Heart J. 2022, 43, 3213–3223. [Google Scholar] [CrossRef]
- Keaney, J.F., Jr. Bempedoic Acid and the Prevention of Cardiovascular Disease. N. Engl. J. Med. 2023, 388, 1427–1430. [Google Scholar] [CrossRef]
- Ray, K.K.; Bays, H.E.; Catapano, A.L.; Lalwani, N.D.; Bloedon, L.T.; Sterling, L.R.; Robinson, P.L.; Ballantyne, C.M.; Trial, C.H. Safety and Efficacy of Bempedoic Acid to Reduce LDL Cholesterol. N. Engl. J. Med. 2019, 380, 1022–1032. [Google Scholar] [CrossRef]
- Nissen, S.E.; Lincoff, A.M.; Brennan, D.; Ray, K.K.; Mason, D.; Kastelein, J.J.P.; Thompson, P.D.; Libby, P.; Cho, L.; Plutzky, J.; et al. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients. N. Engl. J. Med. 2023, 388, 1353–1364. [Google Scholar] [CrossRef]
- Visser, M.E.; Wagener, G.; Baker, B.F.; Geary, R.S.; Donovan, J.M.; Beuers, U.H.; Nederveen, A.J.; Verheij, J.; Trip, M.D.; Basart, D.C.; et al. Mipomersen, an apolipoprotein B synthesis inhibitor, lowers low-density lipoprotein cholesterol in high-risk statin-intolerant patients: A randomized, double-blind, placebo-controlled trial. Eur. Heart J. 2012, 33, 1142–1149. [Google Scholar] [CrossRef] [PubMed]
- Stein, E.A.; Dufour, R.; Gagne, C.; Gaudet, D.; East, C.; Donovan, J.M.; Chin, W.; Tribble, D.L.; McGowan, M. Apolipoprotein B synthesis inhibition with mipomersen in heterozygous familial hypercholesterolemia: Results of a randomized, double-blind, placebo-controlled trial to assess efficacy and safety as add-on therapy in patients with coronary artery disease. Circulation 2012, 126, 2283–2292. [Google Scholar] [CrossRef]
- Santos, R.D.; Raal, F.J.; Catapano, A.L.; Witztum, J.L.; Steinhagen-Thiessen, E.; Tsimikas, S. Mipomersen, an antisense oligonucleotide to apolipoprotein B-100, reduces lipoprotein(a) in various populations with hypercholesterolemia: Results of 4 phase III trials. Arter. Thromb. Vasc. Biol. 2015, 35, 689–699. [Google Scholar] [CrossRef]
- Chambergo-Michilot, D.; Alur, A.; Kulkarni, S.; Agarwala, A. Mipomersen in Familial Hypercholesterolemia: An Update on Health-Related Quality of Life and Patient-Reported Outcomes. Vasc. Health Risk Manag. 2022, 18, 73–80. [Google Scholar] [CrossRef]
- Seidah, N.G.; Prat, A. The Multifaceted Biology of PCSK9. Endocr. Rev. 2022, 43, 558–582. [Google Scholar] [CrossRef]
- Nie, W.; Yue, Y.; Hu, J. PCSK9 inhibitors in the management of atherosclerotic cardiovascular disease: Current clinical trials and future directions. Atherosclerosis 2024, 399, 119043. [Google Scholar] [CrossRef]
- Sabatine, M.S.; Giugliano, R.P.; Keech, A.C.; Honarpour, N.; Wiviott, S.D.; Murphy, S.A.; Kuder, J.F.; Wang, H.; Liu, T.; Wasserman, S.M.; et al. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N. Engl. J. Med. 2017, 376, 1713–1722. [Google Scholar] [CrossRef] [PubMed]
- Schwartz, G.G.; Steg, P.G.; Szarek, M.; Bhatt, D.L.; Bittner, V.A.; Diaz, R.; Edelberg, J.M.; Goodman, S.G.; Hanotin, C.; Harrington, R.A.; et al. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N. Engl. J. Med. 2018, 379, 2097–2107. [Google Scholar] [CrossRef] [PubMed]
- Ray, K.K.; Wright, R.S.; Kallend, D.; Koenig, W.; Leiter, L.A.; Raal, F.J.; Bisch, J.A.; Richardson, T.; Jaros, M.; Wijngaard, P.L.J.; et al. Two Phase 3 Trials of Inclisiran in Patients with Elevated LDL Cholesterol. N. Engl. J. Med. 2020, 382, 1507–1519. [Google Scholar] [CrossRef] [PubMed]
- Natarajan, P.; Kathiresan, S. PCSK9 Inhibitors. Cell 2016, 165, 1037. [Google Scholar] [CrossRef]
- Raal, F.J.; Kallend, D.; Ray, K.K.; Turner, T.; Koenig, W.; Wright, R.S.; Wijngaard, P.L.J.; Curcio, D.; Jaros, M.J.; Leiter, L.A.; et al. Inclisiran for the Treatment of Heterozygous Familial Hypercholesterolemia. N. Engl. J. Med. 2020, 382, 1520–1530. [Google Scholar] [CrossRef]
- Mazer, C.D.; Moody, A.R.; Teoh, H.; Quan, A.; Elituv, R.; Vivekanandan, T.; Li, P.; Thorpe, K.E.; Lafreniere-Roula, M.; Verma, R.; et al. SLICE-CEA CardioLink-8: A Randomized Trial of Evolocumab on Carotid Artery Atherosclerotic Plaque Characteristics in Asymptomatic High-Risk Carotid Stenosis. Circulation 2025, 151, 351–355. [Google Scholar] [CrossRef]
- Bittner, V.A.; Szarek, M.; Aylward, P.E.; Bhatt, D.L.; Diaz, R.; Edelberg, J.M.; Fras, Z.; Goodman, S.G.; Halvorsen, S.; Hanotin, C.; et al. Effect of Alirocumab on Lipoprotein(a) and Cardiovascular Risk After Acute Coronary Syndrome. J. Am. Coll. Cardiol. 2020, 75, 133–144. [Google Scholar] [CrossRef]
- Tsimikas, S. Lipoprotein(a) in the Year 2024: A Look Back and a Look Ahead. Arter. Thromb. Vasc. Biol. 2024, 44, 1485–1490. [Google Scholar] [CrossRef] [PubMed]
- Witztum, J.L.; Gaudet, D.; Freedman, S.D.; Alexander, V.J.; Digenio, A.; Williams, K.R.; Yang, Q.; Hughes, S.G.; Geary, R.S.; Arca, M.; et al. Volanesorsen and Triglyceride Levels in Familial Chylomicronemia Syndrome. N. Engl. J. Med. 2019, 381, 531–542. [Google Scholar] [CrossRef] [PubMed]
- Bergmark, B.A.; Marston, N.A.; Prohaska, T.A.; Alexander, V.J.; Zimerman, A.; Moura, F.A.; Murphy, S.A.; Goodrich, E.L.; Zhang, S.; Gaudet, D.; et al. Olezarsen for Hypertriglyceridemia in Patients at High Cardiovascular Risk. N. Engl. J. Med. 2024, 390, 1770–1780. [Google Scholar] [CrossRef] [PubMed]
- Bergmark, B.A.; Marston, N.A.; Prohaska, T.A.; Alexander, V.J.; Zimerman, A.; Moura, F.A.; Kang, Y.M.; Weinland, J.; Murphy, S.A.; Goodrich, E.L.; et al. Targeting APOC3 with Olezarsen in Moderate Hypertriglyceridemia. N. Engl. J. Med. 2025. [Google Scholar] [CrossRef]
- Nordestgaard, A.T.; Tybjaerg-Hansen, A.; Mansbach, H.; Kersten, S.; Nordestgaard, B.G.; Rosenson, R.S. Target Populations for Novel Triglyceride-Lowering Therapies. J. Am. Coll. Cardiol. 2025, 85, 1876–1897. [Google Scholar] [CrossRef]
- Raal, F.J.; Rosenson, R.S.; Reeskamp, L.F.; Hovingh, G.K.; Kastelein, J.J.P.; Rubba, P.; Ali, S.; Banerjee, P.; Chan, K.C.; Gipe, D.A.; et al. Evinacumab for Homozygous Familial Hypercholesterolemia. N. Engl. J. Med. 2020, 383, 711–720. [Google Scholar] [CrossRef]
- Rosenson, R.S.; Burgess, L.J.; Ebenbichler, C.F.; Baum, S.J.; Stroes, E.S.G.; Ali, S.; Khilla, N.; Hamlin, R.; Pordy, R.; Dong, Y.; et al. Evinacumab in Patients with Refractory Hypercholesterolemia. N. Engl. J. Med. 2020, 383, 2307–2319. [Google Scholar] [CrossRef]
- Rohatgi, A.; Westerterp, M.; von Eckardstein, A.; Remaley, A.; Rye, K.A. HDL in the 21st Century: A Multifunctional Roadmap for Future HDL Research. Circulation 2021, 143, 2293–2309. [Google Scholar] [CrossRef]
- Tall, A.R.; Rader, D.J. Trials and Tribulations of CETP Inhibitors. Circ. Res. 2018, 122, 106–112. [Google Scholar] [CrossRef]
- Armitage, J.; Holmes, M.V.; Preiss, D. Cholesteryl Ester Transfer Protein Inhibition for Preventing Cardiovascular Events: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2019, 73, 477–487. [Google Scholar] [CrossRef] [PubMed]
- Group, H.T.R.C.; Bowman, L.; Hopewell, J.C.; Chen, F.; Wallendszus, K.; Stevens, W.; Collins, R.; Wiviott, S.D.; Cannon, C.P.; Braunwald, E.; et al. Effects of Anacetrapib in Patients with Atherosclerotic Vascular Disease. N. Engl. J. Med. 2017, 377, 1217–1227. [Google Scholar] [CrossRef]
- Krause, B.R.; Remaley, A.T. Reconstituted HDL for the acute treatment of acute coronary syndrome. Curr. Opin. Lipidol. 2013, 24, 480–486. [Google Scholar] [CrossRef] [PubMed]
- Gibson, C.M.; Duffy, D.; Korjian, S.; Bahit, M.C.; Chi, G.; Alexander, J.H.; Lincoff, A.M.; Heise, M.; Tricoci, P.; Deckelbaum, L.I.; et al. Apolipoprotein A1 Infusions and Cardiovascular Outcomes after Acute Myocardial Infarction. N. Engl. J. Med. 2024, 390, 1560–1571. [Google Scholar] [CrossRef] [PubMed]
Circulating Lipids | Normal/Optimal Ranges for Adults |
---|---|
LDL cholesterol | <100 mg/dL (2.6 mM) |
HDL cholesterol | >40 mg/dL (1.0 mM) |
Fasting triglycerides | <150 mg/dL (1.7 mM) |
Lp(a) * | <30 mg/dL (62 nM) |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Steinbeck, J.E.; Iannotti, R.A.; Rasheed, A. Dysregulated Lipid Metabolism as a Central Driver of Atherosclerotic Plaque Pathology. Lipidology 2025, 2, 17. https://doi.org/10.3390/lipidology2040017
Steinbeck JE, Iannotti RA, Rasheed A. Dysregulated Lipid Metabolism as a Central Driver of Atherosclerotic Plaque Pathology. Lipidology. 2025; 2(4):17. https://doi.org/10.3390/lipidology2040017
Chicago/Turabian StyleSteinbeck, Julia Emily, Rachel Anne Iannotti, and Adil Rasheed. 2025. "Dysregulated Lipid Metabolism as a Central Driver of Atherosclerotic Plaque Pathology" Lipidology 2, no. 4: 17. https://doi.org/10.3390/lipidology2040017
APA StyleSteinbeck, J. E., Iannotti, R. A., & Rasheed, A. (2025). Dysregulated Lipid Metabolism as a Central Driver of Atherosclerotic Plaque Pathology. Lipidology, 2(4), 17. https://doi.org/10.3390/lipidology2040017